1
|
Ma L, Gonzalez-Junca A, Chou W, Barcellos-Hoff MH. Monitoring TGFβ signaling in irradiated tumors. Methods Cell Biol 2023; 180:49-67. [PMID: 37890932 DOI: 10.1016/bs.mcb.2023.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2023]
Abstract
Transforming growth factor β (TGFβ) is exquisitely regulated under physiological conditions but its activity is highly dysregulated in cancer. All cells make TGFβ and have receptors for the ligand, which is sequestered in the extracellular matrix in a latent form. Ionizing radiation elicits rapid release of TGFβ from these stores, so-called activation, over a wide range of doses and exposures, including low dose (<1Gy) whole-body irradiation, creating an extraordinarily potent signal in the irradiated tissue or tumor. Hence, accurate evaluation of TGFβ activity is complicated because of its ubiquitous distribution as a latent complex. Here we describe conditions for assays that reveal TGFβ activity in situ using either tissue preparations or functional imaging.
Collapse
Affiliation(s)
- Lin Ma
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, United States
| | - Alba Gonzalez-Junca
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, United States
| | - William Chou
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, United States
| | - Mary Helen Barcellos-Hoff
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, United States.
| |
Collapse
|
2
|
Barcellos-Hoff MH. The radiobiology of TGFβ. Semin Cancer Biol 2022; 86:857-867. [PMID: 35122974 DOI: 10.1016/j.semcancer.2022.02.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 01/27/2023]
Abstract
Ionizing radiation is a pillar of cancer therapy that is deployed in more than half of all malignancies. The therapeutic effect of radiation is attributed to induction of DNA damage that kills cancers cells, but radiation also affects signaling that alters the composition of the tumor microenvironment by activating transforming growth factor β (TGFβ). TGFβ is a ubiquitously expressed cytokine that acts as biological lynchpin to orchestrate phenotypes, the stroma, and immunity in normal tissue; these activities are subverted in cancer to promote malignancy, a permissive tumor microenvironment and immune evasion. The radiobiology of TGFβ unites targets at the forefront of oncology-the DNA damage response and immunotherapy. The cancer cell intrinsic and extrinsic network of TGFβ responses in the irradiated tumor form a barrier to both genotoxic treatments and immunotherapy response. Here, we focus on the mechanisms by which radiation induces TGFβ activation, how TGFβ regulates DNA repair, and the dynamic regulation of the tumor immune microenvironment that together oppose effective cancer therapy. Strategies to inhibit TGFβ exploit fundamental radiobiology that may be the missing link to deploying TGFβ inhibitors for optimal patient benefit from cancer treatment.
Collapse
Affiliation(s)
- Mary Helen Barcellos-Hoff
- Department of Radiation Oncology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
3
|
Hillers-Ziemer LE, Kuziel G, Williams AE, Moore BN, Arendt LM. Breast cancer microenvironment and obesity: challenges for therapy. Cancer Metastasis Rev 2022; 41:627-647. [PMID: 35435599 PMCID: PMC9470689 DOI: 10.1007/s10555-022-10031-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/30/2022] [Indexed: 02/07/2023]
Abstract
Women with obesity who develop breast cancer have a worsened prognosis with diminished survival rates and increased rates of metastasis. Obesity is also associated with decreased breast cancer response to endocrine and chemotherapeutic treatments. Studies utilizing multiple in vivo models of obesity as well as human breast tumors have enhanced our understanding of how obesity alters the breast tumor microenvironment. Changes in the complement and function of adipocytes, adipose-derived stromal cells, immune cells, and endothelial cells and remodeling of the extracellular matrix all contribute to the rapid growth of breast tumors in the context of obesity. Interactions of these cells enhance secretion of cytokines and adipokines as well as local levels of estrogen within the breast tumor microenvironment that promote resistance to multiple therapies. In this review, we will discuss our current understanding of the impact of obesity on the breast tumor microenvironment, how obesity-induced changes in cellular interactions promote resistance to breast cancer treatments, and areas for development of treatment interventions for breast cancer patients with obesity.
Collapse
Affiliation(s)
- Lauren E Hillers-Ziemer
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Genevra Kuziel
- Program in Cancer Biology, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Abbey E Williams
- Comparative Biomedical Sciences Program, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Brittney N Moore
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Lisa M Arendt
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI, 53706, USA.
- Program in Cancer Biology, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Comparative Biomedical Sciences Program, University of Wisconsin-Madison, Madison, WI, 53706, USA.
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, 53706, USA.
- School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Dr. Rm 4354A, Madison, WI, 53706, USA.
| |
Collapse
|
4
|
Dance YW, Meshulam T, Seibel AJ, Obenreder MC, Layne MD, Nelson CM, Tien J. Adipose Stroma Accelerates the Invasion and Escape of Human Breast Cancer Cells from an Engineered Microtumor. Cell Mol Bioeng 2021; 15:15-29. [DOI: 10.1007/s12195-021-00697-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/09/2021] [Indexed: 10/20/2022] Open
|
5
|
Ren G, Rezaee M, Razavi M, Taysir A, Wang J, Thakor AS. Adipose tissue-derived mesenchymal stem cells rescue the function of islets transplanted in sub-therapeutic numbers via their angiogenic properties. Cell Tissue Res 2019; 376:353-364. [PMID: 30707291 DOI: 10.1007/s00441-019-02997-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 01/17/2019] [Indexed: 02/07/2023]
Abstract
A significant proportion of islets are lost following transplantation due to hypoxia and inflammation. We hypothesize that adipose tissue-derived mesenchymal stem cells (AD-MSCs) can rescue a sub-therapeutic number of transplanted islets by helping them establish a new blood supply and reducing inflammation. Diabetic mice received syngeneic transplantation with 75 (minimal), 150 (sub-therapeutic), or 225 (therapeutic) islets, with or without 1 × 106 mouse AD-MSCs. Fasting blood glucose (FBG) values were measured over 6 weeks with tissue samples collected for islet structure and morphology (H&E, insulin/glucagon staining). Histological and immunohistochemical analyses of islets were also performed at 2 weeks in animals transplanted with a sub-therapeutic number of islets, with and without AD-MSCs, to determine new blood vessel formation, the presence of pro-angiogenic factors facilitating revascularization, and the degree of inflammation. AD-MSCs had no beneficial effect on FBG values when co-transplanted with a minimal or therapeutic number of islets. However, AD-MSCs significantly reduced FBG values and restored glycemic control in diabetic animals transplanted with a sub-therapeutic number of islets. Islets co-transplanted with AD-MSCs preserved their native morphology and organization and exhibited less aggregation when compared to islets transplanted alone. In the sub-therapeutic group, AD-MSCs significantly increased islet revascularization and the expression of angiogenic factors including hepatocyte growth factor (HGF) and angiopoietin-1 (Ang-1) while also reducing inflammation. AD-MSCs can rescue the function of islets when transplanted in a sub-therapeutic number, for at least 6 weeks, via their ability to maintain islet architecture while concurrently facilitating islet revascularization and reducing inflammation.
Collapse
Affiliation(s)
- Gang Ren
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University, Department of Radiology, Palo Alto, CA, 94034, USA
| | - Melika Rezaee
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University, Department of Radiology, Palo Alto, CA, 94034, USA.,Chicago Medical School, Rosalind Franklin University, North Chicago, IL, 60064, USA
| | - Mehdi Razavi
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University, Department of Radiology, Palo Alto, CA, 94034, USA
| | - Ahmed Taysir
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University, Department of Radiology, Palo Alto, CA, 94034, USA
| | - Jing Wang
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University, Department of Radiology, Palo Alto, CA, 94034, USA
| | - Avnesh S Thakor
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University, Department of Radiology, Palo Alto, CA, 94034, USA.
| |
Collapse
|
6
|
Ishida K, Mitsui T. Role of the boundary in feather bud formation on one-dimensional bioengineered skin. APL Bioeng 2018; 2:016107. [PMID: 31069292 PMCID: PMC6481706 DOI: 10.1063/1.4989414] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 12/21/2017] [Indexed: 01/04/2023] Open
Abstract
The role of a boundary in pattern formation from a homogenous state in Turing's reaction–diffusion equations is important, particularly when the domain size is comparable to the pattern scale. Such experimental conditions may be achieved for in vitro regeneration of ectodermal appendages such as feathers, via reconstruction of embryonic single cells. This procedure can eliminate a predefined genetic map, such as the midline of chick feather bud formation, leaving uniformly distributed identical cells as a bioengineered skin. Here, the self-organizing nature of multiple feather bud formation was examined in bioengineered 1D-skin samples. Primal formation of feather buds occurred at a fixed length from the skin edge. This formation was numerically recapitulated by a standard two-component reaction-diffusion model, suggesting that the boundary effect caused this observation. The proper boundary conditions were nonstandard, either mixed Dirichlet–Neumann or partial-flux. In addition, the model implies imperfect or hindered bud formation as well as nearly equal distances between buds. In contrast, experimental observations indicated that the skin curvature, which was not included in our model, also strongly affected bud formation. Thus, bioengineered skin may provide an ideal template for modeling a self-organized process from a homogenous state. This study will examine the possible diffusion activities of activator or inhibitor molecular candidates and mechanical activities during cell aggregation, which will advance our understanding of skin appendage regeneration from pluripotent or embryonic stem cells.
Collapse
Affiliation(s)
- Kentaro Ishida
- Department of Physics and Mathematics, College of Science and Engineering, Aoyama Gakuin University, Kanagawa 252-5258, Japan
| | - Toshiyuki Mitsui
- Department of Physics and Mathematics, College of Science and Engineering, Aoyama Gakuin University, Kanagawa 252-5258, Japan
| |
Collapse
|
7
|
Fibronectin, the extracellular glue. Matrix Biol 2017; 60-61:27-37. [DOI: 10.1016/j.matbio.2016.07.011] [Citation(s) in RCA: 180] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 07/22/2016] [Accepted: 07/30/2016] [Indexed: 12/13/2022]
|
8
|
Barbosa GO, Cervigne NK, Carvalho HF, Augusto TM. Heparanase 1 involvement in prostate physiopathology. Cell Biol Int 2017; 41:1194-1202. [DOI: 10.1002/cbin.10748] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 02/13/2017] [Indexed: 12/25/2022]
Affiliation(s)
- Guilherme O. Barbosa
- Department of Structural and Functional Biology; State University of Campinas; Campinas Sao Paulo Brazil
| | - Nilva K. Cervigne
- Faculty of Medicine of Jundiai; Department of Morphology and Basic Pathology; Jundiai Sao Paulo Brazil
| | - Hernandes F. Carvalho
- Department of Structural and Functional Biology; State University of Campinas; Campinas Sao Paulo Brazil
| | - Taize M. Augusto
- Faculty of Medicine of Jundiai; Department of Morphology and Basic Pathology; Jundiai Sao Paulo Brazil
| |
Collapse
|
9
|
Fercana GR, Yerneni S, Billaud M, Hill JC, VanRyzin P, Richards TD, Sicari BM, Johnson SA, Badylak SF, Campbell PG, Gleason TG, Phillippi JA. Perivascular extracellular matrix hydrogels mimic native matrix microarchitecture and promote angiogenesis via basic fibroblast growth factor. Biomaterials 2017; 123:142-154. [PMID: 28167392 DOI: 10.1016/j.biomaterials.2017.01.037] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 01/27/2017] [Indexed: 01/12/2023]
Abstract
Extracellular matrix (ECM)-derived bioscaffolds have been shown to elicit tissue repair through retention of bioactive signals. Given that the adventitia of large blood vessels is a richly vascularized microenvironment, we hypothesized that perivascular ECM contains bioactive signals that influence cells of blood vessel lineages. ECM bioscaffolds were derived from decellularized human and porcine aortic adventitia (hAdv and pAdv, respectively) and then shown have minimal DNA content and retain elastin and collagen proteins. Hydrogel formulations of hAdv and pAdv ECM bioscaffolds exhibited gelation kinetics similar to ECM hydrogels derived from porcine small intestinal submucosa (pSIS). hAdv and pAdv ECM hydrogels displayed thinner, less undulated, and fibrous microarchitecture reminiscent of native adventitia, with slight differences in ultrastructure visible in comparison to pSIS ECM hydrogels. Pepsin-digested pAdv and pSIS ECM bioscaffolds increased proliferation of human adventitia-derived endothelial cells and this effect was mediated in part by basic fibroblast growth factor (FGF2). Human endothelial cells cultured on Matrigel substrates formed more numerous and longer tube-like structures when supplemented with pAdv ECM bioscaffolds, and FGF2 mediated this matrix signaling. ECM bioscaffolds derived from pAdv promoted FGF2-dependent in vivo angiogenesis in the chick chorioallantoic membrane model. Using an angiogenesis-focused protein array, we detected 55 angiogenesis-related proteins, including FGF2 in hAdv, pAdv and pSIS ECMs. Interestingly, 19 of these factors were less abundant in ECMs bioscaffolds derived from aneurysmal specimens of human aorta when compared with non-aneurysmal (normal) specimens. This study reveals that Adv ECM hydrogels recapitulate matrix fiber microarchitecture of native adventitia, and retain angiogenesis-related actors and bioactive properties such as FGF2 signaling capable of influencing processes important for angiogenesis. This work supports the use of Adv ECM bioscaffolds for both discovery biology and potential translation towards microvascular regeneration in clinical applications.
Collapse
Affiliation(s)
- George R Fercana
- Department of Cardiothoracic Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Saigopalakrishna Yerneni
- Department of Cardiothoracic Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Marie Billaud
- Department of Cardiothoracic Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Jennifer C Hill
- Department of Cardiothoracic Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Paul VanRyzin
- Department of Cardiothoracic Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Tara D Richards
- Department of Cardiothoracic Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Brian M Sicari
- Department of Cardiothoracic Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Scott A Johnson
- Department of Cardiothoracic Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Stephen F Badylak
- Department of Cardiothoracic Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Phil G Campbell
- Department of Cardiothoracic Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Thomas G Gleason
- Department of Cardiothoracic Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Julie A Phillippi
- Department of Cardiothoracic Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
10
|
Freudenberg U, Zieris A, Chwalek K, Tsurkan MV, Maitz MF, Atallah P, Levental KR, Eming SA, Werner C. Heparin desulfation modulates VEGF release and angiogenesis in diabetic wounds. J Control Release 2015; 220:79-88. [DOI: 10.1016/j.jconrel.2015.10.028] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 09/28/2015] [Accepted: 10/14/2015] [Indexed: 10/22/2022]
|
11
|
Tanaka M, Yamaguchi M, Shiota M, Kawamoto Y, Takahashi K, Inagaki A, Osada-Oka M, Harada A, Wanibuchi H, Izumi Y, Miura K, Iwao H, Ohkawa Y. Establishment of neutralizing rat monoclonal antibodies for fibroblast growth factor-2. Monoclon Antib Immunodiagn Immunother 2015; 33:261-9. [PMID: 25171006 DOI: 10.1089/mab.2013.0085] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Fibroblast growth factor-2 (FGF-2) plays a critical role in endothelial survival, proliferation, and angiogenesis and is localized on the cell membrane by binding to heparan sulfate proteoglycans. Here we established a neutralizing monoclonal antibody, 1B9B9, against FGF-2 using the rat medial iliac lymph node method. 1B9B9 blocked the binding of FGF-2 to its receptor, inhibiting FGF-2-induced proliferation and corresponding downstream signaling in endothelial cells. Treatment of human umbilical vein endothelial cells with 1B9B9 reduced the basal phosphorylation levels of Akt and MAPK. Furthermore, continued treatment with 1B9B9 induced cell death by apoptosis. Compared with FGF-2 knockdown, 1B9B9 significantly reduced cell survival. In addition, the combination of FGF-2 siRNA and 1B9B9 showed a synergistic effect. The data indicate that 1B9B9 established by the rat iliac lymph node method is a fully compatible neutralizing antibody.
Collapse
Affiliation(s)
- Masako Tanaka
- 1 Department of Pharmacology, Osaka City University Medical School , Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Pellicciotta I, Marciscano AE, Hardee ME, Francis D, Formenti S, Barcellos-Hoff MH. Development of a novel multiplexed assay for quantification of transforming growth factor-β (TGF-β). Growth Factors 2015; 33:79-91. [PMID: 25586866 DOI: 10.3109/08977194.2014.999367] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Changes in activity or levels of transforming growth factor-β (TGF-β) are associated with a variety of diseases; however, measurement of TGF-β in biological fluids is highly variable. TGF-β is biologically inert when associated with its latency-associated peptide (LAP). Most available immunoassays require exogenous activation by acid/heat to release TGF-β from the latent complex. We developed a novel electrochemiluminescence-based multiplexed assay on the MesoScale Discovery® platform that eliminates artificial activation, simultaneously measures both active TGF-β1 and LAP1 and includes an internal control for platelet-derived TGF-β contamination in blood specimens. We optimized this assay to evaluate plasma levels as a function of activation type and clinical specimen preparation. We determined that breast cancer patients' plasma have higher levels of circulating latent TGF-β (LTGF-β) as measured by LAP1 than healthy volunteers (p < 0.0001). This assay provides a robust tool for correlative studies of LTGF-β levels with disease, treatment outcomes and toxicity with a broad clinical applicability.
Collapse
|
13
|
Mutant MMP-9 and HGF gene transfer enhance resolution of CCl4-induced liver fibrosis in rats: role of ASH1 and EZH2 methyltransferases repression. PLoS One 2014; 9:e112384. [PMID: 25380300 PMCID: PMC4224431 DOI: 10.1371/journal.pone.0112384] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 10/03/2014] [Indexed: 12/20/2022] Open
Abstract
Hepatocyte growth factor (HGF) gene transfer inhibits liver fibrosis by regulating aberrant cellular functions, while mutant matrix metalloproteinase-9 (mMMP-9) enhances matrix degradation by neutralizing the elevated tissue inhibitor of metalloproteinase-1 (TIMP-1). It was shown that ASH1 and EZH2 methyltransferases are involved in development of liver fibrosis; however, their role in the resolution phase of liver fibrosis has not been investigated. This study evaluated the role of ASH1 and EZH2 in two mechanistically different therapeutic modalities, HGF and mMMP-9 gene transfer in CCl4 induced rat liver fibrosis. Liver fibrosis was induced in rats with twice a week intraperitoneal injection of CCl4 for 8 weeks. Adenovirus vectors encoding mMMP-9 or HGF genes were injected through tail vein at weeks six and seven and were sacrificed one week after the second injection. A healthy animal group was likewise injected with saline to serve as a negative control. Rats treated with mMMP-9 showed significantly lower fibrosis score, less Sirius red stained collagen area, reduced hydroxyproline and ALT concentration, decreased transforming growth factor beta 1 (TGF-β1) mRNA and lower labeling indices of α smooth muscle actin (α-SMA) and proliferating cell nuclear antigen (PCNA) stained cells compared with HGF- or saline-treated rats. Furthermore, TIMP-1 protein expression in mMMP-9 group was markedly reduced compared with all fibrotic groups. ASH1 and EZH2 protein expression was significantly elevated in fibrotic liver and significantly decreased in mMMP-9- and HGF-treated compared to saline-treated fibrotic livers with further reduction in the mMMP-9 group. Conclusion: Gene transfer of mMMP-9 and HGF reduced liver fibrosis in rats. ASH1 and EZH2 methyltransferases are significantly reduced in mMMP-9 and HGF treated rats which underlines the central role of these enzymes during fibrogenesis. Future studies should evaluate the role of selective pharmacologic inhibitors of ASH1 and EZH2 in resolution of liver fibrosis.
Collapse
|
14
|
Vivès RR, Seffouh A, Lortat-Jacob H. Post-Synthetic Regulation of HS Structure: The Yin and Yang of the Sulfs in Cancer. Front Oncol 2014; 3:331. [PMID: 24459635 PMCID: PMC3890690 DOI: 10.3389/fonc.2013.00331] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 12/27/2013] [Indexed: 12/11/2022] Open
Abstract
Heparan sulfate (HS) is a complex polysaccharide that takes part in most major cellular processes, through its ability to bind and modulate a very large array of proteins. These interactions involve saccharide domains of specific sulfation pattern (S-domains), the assembly of which is tightly orchestrated by a highly regulated biosynthesis machinery. Another level of structural control does also take place at the cell surface, where degrading enzymes further modify HS post-synthetically. Amongst them are the Sulfs, a family of extracellular sulfatases (two isoforms in human) that catalyze the specific 6-O-desulfation of HS. By targeting HS functional sulfated domains, Sulfs dramatically alter its ligand binding properties, thereby modulating a broad range of signaling pathways. Consequently, Sulfs play major roles during development, as well as in tissue homeostasis and repair. Sulfs have also been associated with many pathologies including cancer, but despite increasing interest, the role of Sulfs in tumor development still remains unclear. Studies have been hindered by a poor understanding of the Sulf enzymatic activities and conflicting data have shown either anti-oncogenic or tumor-promoting effects of these enzymes, depending on the tumor models analyzed. These opposite effects clearly illustrate the fine tuning of HS functions by the Sulfs, and the need to clarify the mechanisms involved. In this review, we will detail the present knowledge on the structural and functional properties of the Sulfs, with a special focus on their implication during tumor progression. Finally, we will discuss attempts and perspectives of using the Sulfs as a biomarker of cancer prognosis and diagnostic and as a target for anti-cancer therapies.
Collapse
Affiliation(s)
- Romain R Vivès
- Université Grenoble-Alpes, Institut de Biologie Structurale , Grenoble , France ; CNRS, Institut de Biologie Structurale , Grenoble , France ; CEA, DSV, Institut de Biologie Structurale , Grenoble , France
| | - Amal Seffouh
- Université Grenoble-Alpes, Institut de Biologie Structurale , Grenoble , France ; CNRS, Institut de Biologie Structurale , Grenoble , France ; CEA, DSV, Institut de Biologie Structurale , Grenoble , France
| | - Hugues Lortat-Jacob
- Université Grenoble-Alpes, Institut de Biologie Structurale , Grenoble , France ; CNRS, Institut de Biologie Structurale , Grenoble , France ; CEA, DSV, Institut de Biologie Structurale , Grenoble , France
| |
Collapse
|
15
|
Abstract
Receptors for basic (b) and acidic (a) fibroblast growth factor (FGF) are upregulated in activated smooth muscle cells. These cells, which proliferate in response to bFGF, can thus be killed by a conjugate of bFGF and the ribosome-inactivating enzyme, saporin (which, by itself, does not enter the cells). Quiescent smooth muscle cells and other cells that have few FGF receptors are not killed. In vivo, bFGF-saporin transiently inhibits smooth muscle cell proliferation and neointimal accumulation after balloon injury to the rat carotid artery. Delivery of saporin, diagnostic imaging agents, or antisense oligodeoxynucleotides might be made even more selective by linking these substances to antibodies against the extracellular domains of the putative FGF receptor isoform specific for activated smooth muscle cells.
Collapse
Affiliation(s)
- W Casscells
- Texas Heart Institute and University of Texas Medical School, Houston, TX 77225-0345 USA
| | | | | |
Collapse
|
16
|
Transforming growth factor (TGF)-β expression and activation mechanisms as potential targets for anti-tumor therapy and tumor imaging. Pharmacol Ther 2012; 135:123-32. [DOI: 10.1016/j.pharmthera.2012.05.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Accepted: 04/10/2012] [Indexed: 11/23/2022]
|
17
|
Choi I, Yeo WS. Self-Assembled Monolayers with Dynamicity Stemming from (Bio)Chemical Conversions: From Construction to Application. Chemphyschem 2012; 14:55-69. [DOI: 10.1002/cphc.201200293] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Indexed: 11/11/2022]
|
18
|
Mathews S, Mathew SA, Gupta PK, Bhonde R, Totey S. Glycosaminoglycans enhance osteoblast differentiation of bone marrow derived human mesenchymal stem cells. J Tissue Eng Regen Med 2012; 8:143-52. [PMID: 22499338 DOI: 10.1002/term.1507] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Revised: 12/09/2011] [Accepted: 02/01/2012] [Indexed: 01/29/2023]
Abstract
Extracellular matrix plays an important role in regulating cell growth and differentiation. The biomimetic approach of cell-based tissue engineering is based on mirroring this in vivo micro environment for developing a functional tissue engineered construct. In this study, we treated normal tissue culture plates with selected extracellular matrix components consisting of glycosaminoglycans such as chondroitin-4-sulphate, dermatan sulphate, chondroitin-6-sulphate, heparin and hyaluronic acid. Mesenchymal stem cells isolated from adult human bone marrow were cultured on the glycosaminoglycan treated culture plates to evaluate their regulatory role in cell growth and osteoblast differentiation. Although no significant improvement on human mesenchymal stem cell adhesion and proliferation was observed on the glycosaminoglycan-treated tissue culture plates, there was selective osteoblast differentiation, indicating its potential role in differentiation rather than proliferation. Osteoblast differentiation studies showed high osteogenic potential for all tested glycosaminoglycans except chondroitin-4-sulphate. Osteoblast differentiation-associated genes such as osterix, osteocalcin, integrin binding sialoprotein, osteonectin and collagen, type 1, alpha 1 showed significant upregulation. We identified osterix as the key transcription factor responsible for the enhanced bone matrix deposition observed on hyaluronic acid, heparin and chondroitin-6-sulphate. Hyaluronic acid provided the most favourable condition for osteoblast differentiation and bone matrix synthesis. Our results confirm and emphasise the significant role of extracellular matrix in regulating cell differentiation. To summarise, glycosaminoglycans of extracellular matrix played a significant role in regulating osteoblast differentiation and could be exploited in the biomimetic approach of fabricating or functionalizing scaffolds for stem cell based bone tissue engineering.
Collapse
Affiliation(s)
- Smitha Mathews
- Manipal Institute of Regenerative Medicine, Manipal University, 10 Service Road, Domlur, Bangalore-560071, India
| | | | | | | | | |
Collapse
|
19
|
Zieris A, Prokoph S, Levental KR, Welzel PB, Chwalek K, Schneider K, Freudenberg U, Werner C. Sustainable Growth Factor Delivery through Affinity-Based Adsorption to starPEG-Heparin Hydrogels. ACS SYMPOSIUM SERIES 2012. [DOI: 10.1021/bk-2012-1120.ch024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
|
20
|
Nagaraju GPC, Sharma D. Anti-cancer role of SPARC, an inhibitor of adipogenesis. Cancer Treat Rev 2011; 37:559-66. [PMID: 21237573 PMCID: PMC3139810 DOI: 10.1016/j.ctrv.2010.12.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 12/01/2010] [Accepted: 12/07/2010] [Indexed: 12/17/2022]
Abstract
SPARC (a secreted protein acidic and rich in cysteine) has a reputation for being potent anti-cancer and anti-obesity molecule. It is one of the first known matricellular protein that modulates interactions between cells and extracellular matrix (ECM) and is associated with the 'balance' of white adipose tissue (WAT) as well as lipogenesis and lipolysis during adipogenesis. Adipogenesis is an indication for the development of obesity and has been related to a wide variety of cancers including breast cancer, endometrial cancer, esophageal cancer, etc. Adipogenesis mainly involves ECM remodeling, changes in cell-ECM interactions, and cytoskeletal rearrangement. SPARC can also prevent hypertrophy of adipocytes and hyperplasia of adipocyte progenitors. In addition to SPARC's inhibitory role in adipogenesis, it has also been known to be involved in cell cycle, cell proliferation, cell invasion, adhesion, migration, angiogenesis and apoptosis. Molecular cancer biology and clinical biochemistry have significantly enhanced our understanding of the mechanisms that motivate the anti-cancer and anti-obesity action of SPARC. Recent studies elucidating the signaling pathways that are activated by SPARC can help develop the beneficial aspects of SPARC for cancer therapy and obesity prevention. This review focuses on the anti-cancer role of SPARC as it pertains to obesity.
Collapse
Affiliation(s)
| | - Dipali Sharma
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta GA 30322
- Emory Winship Cancer Institute, Emory University School of Medicine, Atlanta GA 30322
| |
Collapse
|
21
|
Zieris A, Chwalek K, Prokoph S, Levental K, Welzel P, Freudenberg U, Werner C. Dual independent delivery of pro-angiogenic growth factors from starPEG-heparin hydrogels. J Control Release 2011; 156:28-36. [DOI: 10.1016/j.jconrel.2011.06.042] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Revised: 06/23/2011] [Accepted: 06/28/2011] [Indexed: 10/18/2022]
|
22
|
Sullivan LA, Brekken RA. The VEGF family in cancer and antibody-based strategies for their inhibition. MAbs 2011; 2:165-75. [PMID: 20190566 DOI: 10.4161/mabs.2.2.11360] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Angiogenesis is required in normal physiological processes, but is also involved in tumor growth, progression and metastasis. Vascular endothelial growth factor (VEGF), a primary mediator of angiogenesis in normal physiology and in disease, and other VEGF family members and their receptors provide targets that have been explored extensively for cancer therapy. Small molecule inhibitors and antibody/protein-based strategies that target the VEGF pathway have been studied in multiple types of cancer. This review will focus on VEGF pathway targeting antibodies that are currently being evaluated in pre-clinical and clinical studies.
Collapse
Affiliation(s)
- Laura A Sullivan
- Division of Surgical Oncology, Department of Surgery, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, USA
| | | |
Collapse
|
23
|
Nguyen DH, Martinez-Ruiz H, Barcellos-Hoff MH. Consequences of epithelial or stromal TGFβ1 depletion in the mammary gland. J Mammary Gland Biol Neoplasia 2011; 16:147-55. [PMID: 21590374 DOI: 10.1007/s10911-011-9218-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Accepted: 03/14/2011] [Indexed: 01/21/2023] Open
Abstract
Transforming growth factor β1 (TGFβ) affects stroma and epithelial composition and interactions that mediate mammary development and determine the course of cancer. The reduction of TGFβ in Tgfβ1 heterozygote mice, which are healthy and long-lived, provides an important model to dissect the contribution of TGFβ in mammary gland biology and cancer. We used both intact mice and mammary chimeras in conjunction with Tgfβ1 genetic depletion and TGFβ neutralizing antibodies to evaluate how stromal or epithelial TGFβ depletion affect mammary development and response to physiological stimuli. Our studies of radiation carcinogenesis have revealed new aspects of TGFβ biology and suggest that the paradoxical TGFβ switch from tumor suppressor to tumor promoter can be resolved by assessing distinct stromal versus epithelial actions.
Collapse
Affiliation(s)
- David H Nguyen
- Endocrinology Graduate Group, University of California, Berkeley, CA, USA
| | | | | |
Collapse
|
24
|
Moses H, Barcellos-Hoff MH. TGF-beta biology in mammary development and breast cancer. Cold Spring Harb Perspect Biol 2011; 3:a003277. [PMID: 20810549 DOI: 10.1101/cshperspect.a003277] [Citation(s) in RCA: 190] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Transforming growth factor-β1 (TGF-β) was first implicated in mammary epithelial development by Daniel and Silberstein in 1987 and in breast cancer cells and hormone resistance by Lippman and colleagues in 1988. TGF-β is critically important for mammary morphogenesis and secretory function through specific regulation of epithelial proliferation, apoptosis, and extracellular matrix. Differential TGF-β effects on distinct cell types are compounded by regulation at multiple levels and the influence of context on cellular responses. Studies using controlled expression and conditional-deletion mouse models underscore the complexity of TGF-β biology across the cycle of mammary development and differentiation. Early loss of TGF-β growth regulation in breast cancer evolves into fundamental deregulation that mediates cell interactions and phenotypes driving invasive disease. Two outstanding issues are to understand the mechanisms of biological control in situ and the circumstances by which TGF-β regulation is subverted in neoplastic progression.
Collapse
Affiliation(s)
- Harold Moses
- Department of Cancer Biology and Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee 37232, USA
| | | |
Collapse
|
25
|
Fukui-Miyazaki A, Kamitani S, Miyake M, Horiguchi Y. Association of Bordetella dermonecrotic toxin with the extracellular matrix. BMC Microbiol 2010; 10:247. [PMID: 20868510 PMCID: PMC2949704 DOI: 10.1186/1471-2180-10-247] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Accepted: 09/25/2010] [Indexed: 11/12/2022] Open
Abstract
Background Bordetella dermonecrotic toxin (DNT) causes the turbinate atrophy in swine atrophic rhinitis, caused by a Bordetella bronchiseptica infection of pigs, by inhibiting osteoblastic differentiation. The toxin is not actively secreted from the bacteria, and is presumed to be present in only small amounts in infected areas. How such small amounts can affect target tissues is unknown. Results Fluorescence microscopy revealed that DNT associated with a fibrillar structure developed on cultured cells. A cellular component cross-linked with DNT conjugated with a cross-linker was identified as fibronectin by mass spectrometry. Colocalization of the fibronectin network on the cells with DNT was also observed by fluorescence microscope. Several lines of evidence suggested that DNT interacts with fibronectin not directly, but through another cellular component that remains to be identified. The colocalization was observed in not only DNT-sensitive cells but also insensitive cells, indicating that the fibronectin network neither serves as a receptor for the toxin nor is involved in the intoxicating procedures. The fibronectin network-associated toxin was easily liberated when the concentration of toxin in the local environment decreased, and was still active. Conclusions Components in the extracellular matrix are known to regulate activities of various growth factors by binding and liberating them in response to alterations in the extracellular environment. Similarly, the fibronectin-based extracellular matrix may function as a temporary storage system for DNT, enabling small amounts of the toxin to efficiently affect target tissues or cells.
Collapse
Affiliation(s)
- Aya Fukui-Miyazaki
- Department of Molecular Bacteriology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.
| | | | | | | |
Collapse
|
26
|
Barcellos-Hoff MH, Newcomb EW, Zagzag D, Narayana A. Therapeutic targets in malignant glioblastoma microenvironment. Semin Radiat Oncol 2009; 19:163-70. [PMID: 19464631 DOI: 10.1016/j.semradonc.2009.02.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
There is considerable evidence that the tissue microenvironment can suppress cancer and that microenvironment disruption is required for cancer growth and progression. Distortion of the microenvironment by tumor cells can promote growth, recruit nonmalignant cells that provide physiological resources, and facilitate invasion. Compared with the variable routes taken by cells to become cancers, the response of normal tissue to cancer is relatively consistent such that controlling cancer may be more readily achieved indirectly via the microenvironment. Here, we discuss 3 ideas about how the microenvironment, consisting of a vasculature, inflammatory cells, immune cells, growth factors, and extracellular matrix, might provide therapeutic targets in glioblastoma (GBM) in the context of radiotherapy (RT): (1) viable therapeutic targets exist in the GBM microenvironment, (2) RT alters the microenvironment of tissues and tumors; and (3) a potential benefit may be achieved by targeting the microenvironments induced by RT.
Collapse
Affiliation(s)
- Mary Helen Barcellos-Hoff
- Department of Radiation Oncology, New York University, Langone School of Medicine, New York, NY 10016, USA.
| | | | | | | |
Collapse
|
27
|
Esposito C, Parrilla B, Cornacchia F, Grosjean F, Mangione F, Serpieri N, Valentino R, Villa L, Arra M, Esposito V, Dal Canton A. The antifibrogenic effect of hepatocyte growth factor (HGF) on renal tubular (HK-2) cells is dependent on cell growth. Growth Factors 2009; 27:173-80. [PMID: 19301208 DOI: 10.1080/08977190902834077] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Although several reports suggest an antifibrogenic effect of hepatocyte growth factor (HGF), an increased deposition of matrix induced by HGF has also been reported. These conflicting effects could result from a diverse proliferative state of the target cells. Aim of the present study was to evaluate HGF effects on growth arrested (quiescent) and actively proliferating renal tubular epithelial (HK-2) cells. HK-2 cells were cultured in RPMI medium either on agarose gel or on plastic surface in order to inhibit or to allow cell proliferation. Cells were incubated with RPMI containing HGF (50 ng/ml) for 24 h at 37 degrees C. Untreated HK-2 were used as control. After 24 h of incubation, cells were counted by Coulter counter. (alpha2)IV collagen, transforming growth factor-beta (TGF-beta), Tissue inhibitor of metalloproteases (TIMP1 and 2) mRNA levels were determined by RT-PCR. The production of type IV collagen, c-met, proliferating cell nuclear antigen (PCNA), and SnoN, a transcriptional Smad corepressor and thus a TGF-beta inhibitor, was evaluated by ELISA or western blotting. MMP-9 and 2 gelatinolytic activity was studied by zymography. Treatment with HGF did not increase HK-2 cell number and PCNA synthesis when the cells were grown on agarose as it did for cells grown on plastic surface. HGF increased (alpha2)IV collagen in proliferating cells whereas it reduced (alpha2)IV collagen and c-met synthesis in growth arrested cells. HGF treatment increased TGF-beta and TIMP-2 in proliferating cells while reduced TIMP-1 mRNA levels of quiescent cells. Furthermore, production of the co repressor SnoN was significantly decreased by HGF in proliferating cells. Quiescent and proliferating HK-2 showed a different pattern of metalloproteases activity with a prevalence of MMP2 in quiescent and MMP9 in proliferating cells. In summary, HGF showed opposite effects on growth arrested and proliferating HK-2 cells favouring matrix deposition in the latter with increasing expression of collagen, TIMP-1 and TGF-beta. Our results demonstrate that the proliferative state of target cells may influence the effects of HGF on extracellular matrix turnover in HK-2 cells.
Collapse
Affiliation(s)
- Ciro Esposito
- Unit of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Stendahl JC, Kaufman DB, Stupp SI. Extracellular matrix in pancreatic islets: relevance to scaffold design and transplantation. Cell Transplant 2009; 18:1-12. [PMID: 19476204 PMCID: PMC2724969 DOI: 10.3727/096368909788237195] [Citation(s) in RCA: 254] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Intrahepatic islet transplantation provides a potentially more benign alternative to pancreatic transplantation. However, islet transplants are associated with limited engraftment potential. This inefficiency is likely at least partially attributable to the isolation process, which removes islets from their native environment. Isolation not only disrupts the internal vascularization and innervation of islets, but also fundamentally changes interactions between islet cells and macromolecules of the extracellular matrix (ECM). Signaling interactions between islet cells and ECM are known to regulate multiple aspects of islet physiology, including survival, proliferation, and insulin secretion. Although it is highly likely that disruptions to these interactions during isolation significantly affect transplant outcomes, the true implications of these conditions are not well understood. The following article reviews current understandings and uncertainties in islet-ECM interactions and explains their potential impact on posttransplant engraftment. Topics covered include matrix and receptor compositions in native islets, effects of isolation and culture on islet-ECM interactions, and potential for postisolation restoration of islet-ECM interactions. Greater understanding in these areas may help to reduce isolation and transplantation stresses and improve islet engraftment.
Collapse
Affiliation(s)
- John C. Stendahl
- Institute for BioNanotechnology in Advanced Medicine, Northwestern University, Chicago, IL, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
| | - Dixon B. Kaufman
- Institute for BioNanotechnology in Advanced Medicine, Northwestern University, Chicago, IL, USA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Samuel I. Stupp
- Institute for BioNanotechnology in Advanced Medicine, Northwestern University, Chicago, IL, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
- Department of Chemistry, Northwestern University, Evanston, IL, USA
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
29
|
Abstract
Fibrosis is characterized by excessive accumulation of extracellular matrix (ECM) in basement membranes and interstitial tissues, resulting from increased synthesis or decreased degradation of ECM or both. The plasminogen activator/plasmin system plays an important role in ECM degradation, whereas the plasminogen activator inhibitor 1 (PAI-1) is a physiologic inhibitor of plasminogen activators. PAI-1 expression is increased in the lung fibrotic diseases and in experimental fibrosis models. The deletion of the PAI-1 gene reduces, whereas the overexpression of PAI-1 enhances, the susceptibility of animals to lung fibrosis induced by different stimuli, indicating an important role of PAI-1 in the development of lung fibrosis. Many growth factors, including transforming growth factor beta (TGF-beta) and tumor necrosis factor alpha (TNF-alpha), as well as other chemicals/agents, induce PAI-1 expression in cultured cells and in vivo. Reactive oxygen and nitrogen species (ROS/RNS) have been shown to mediate the induction of PAI-1 by many of these stimuli. This review summarizes some recent findings that help us to understand the role of PAI-1 in the development of lung fibrosis and ROS/RNS in the regulation of PAI-1 expression during fibrogenesis.
Collapse
Affiliation(s)
- Rui-Ming Liu
- Department of Environmental Health Sciences, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA.
| |
Collapse
|
30
|
Biondi M, Ungaro F, Quaglia F, Netti PA. Controlled drug delivery in tissue engineering. Adv Drug Deliv Rev 2008; 60:229-42. [PMID: 18031864 DOI: 10.1016/j.addr.2007.08.038] [Citation(s) in RCA: 285] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2007] [Accepted: 08/09/2007] [Indexed: 11/15/2022]
Abstract
The concept of tissue and cell guidance is rapidly evolving as more information regarding the effect of the microenvironment on cellular function and tissue morphogenesis become available. These disclosures have lead to a tremendous advancement in the design of a new generation of multifunctional biomaterials able to mimic the molecular regulatory characteristics and the three-dimensional architecture of the native extracellular matrix. Micro- and nano-structured scaffolds able to sequester and deliver in a highly specific manner biomolecular moieties have already been proved to be effective in bone repairing, in guiding functional angiogenesis and in controlling stem cell differentiation. Although these platforms represent a first attempt to mimic the complex temporal and spatial microenvironment presented in vivo, an increased symbiosis of material engineering, drug delivery technology and cell and molecular biology may ultimately lead to biomaterials that encode the necessary signals to guide and control developmental process in tissue- and organ-specific differentiation and morphogenesis.
Collapse
Affiliation(s)
- Marco Biondi
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Naples, Italy
| | | | | | | |
Collapse
|
31
|
Roskoski R. Vascular endothelial growth factor (VEGF) signaling in tumor progression. Crit Rev Oncol Hematol 2007; 62:179-213. [PMID: 17324579 DOI: 10.1016/j.critrevonc.2007.01.006] [Citation(s) in RCA: 439] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2006] [Revised: 01/01/2007] [Accepted: 01/29/2007] [Indexed: 12/17/2022] Open
Abstract
Vascular endothelial cells are ordinarily quiescent in adult humans and divide less than once per decade. When tumors reach a size of about 0.2-2.0mm in diameter, they become hypoxic and limited in size in the absence of angiogenesis. There are about 30 endogenous pro-angiogenic factors and about 30 endogenous anti-angiogenic factors. In order to increase in size, tumors undergo an angiogenic switch where the action of pro-angiogenic factors predominates, resulting in angiogenesis and tumor progression. One mechanism for driving angiogenesis results from the increased production of vascular endothelial growth factor (VEGF) following up-regulation of the hypoxia-inducible transcription factor. The human VEGF family consists of VEGF (VEGF-A), VEGF-B, VEGF-C, VEGF-D, and placental growth factor (PlGF). The VEGF family of receptors consists of three protein-tyrosine kinases and two non-protein kinase receptors (neuropilin-1 and -2). Owing to the importance of angiogenesis in tumor progression, inhibition of VEGF signaling represents an attractive cancer treatment.
Collapse
Affiliation(s)
- Robert Roskoski
- Blue Ridge Institute for Medical Research, 3754 Brevard Road, Suite 116A, Box 19, Horse Shoe, NC 28742, USA.
| |
Collapse
|
32
|
The Fibroblast Growth Factor (FGF) – FGF Receptor Complex: Progress Towards the Physiological State. Top Curr Chem (Cham) 2006. [DOI: 10.1007/128_068] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
33
|
Wollenweber M, Domaschke H, Hanke T, Boxberger S, Schmack G, Gliesche K, Scharnweber D, Worch H. Mimicked bioartificial matrix containing chondroitin sulphate on a textile scaffold of poly(3-hydroxybutyrate) alters the differentiation of adult human mesenchymal stem cells. ACTA ACUST UNITED AC 2006; 12:345-59. [PMID: 16548693 DOI: 10.1089/ten.2006.12.345] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Controlling the differentiation of human mesenchymal stem cells (hMSC) and providing tissue functions in engineered constructs before implantation are major challenges. Beside the additives in culture media, the artificial niches inside a scaffold can serve this purpose. To prepare niches favoring the osteoblastic differentiation of hMSCs, components of the extracellular matrix of bone were immobilized on fabrics of poly(3-hydroxybutyrate). Aqueous gels of fibrillar bovine collagen I, with or without addition of chondroitin sulphate (CS), were immobilized on the textile scaffold, sub-structured in a freeze-drying process, and cross-linked. hMSCs of four donors were isolated from bone marrow. After expansion, the cells were seeded dynamically onto the scaffolds. From thereon, the culture was transferred into perfused vessels and partly submitted to dexamethasone to promote osteogenic differentiation. During their 4 weeks of culture, the cells' distribution and morphology throughout the scaffolds were characterized by laser scanning microscopy (LSM) and scanning electron microscopy (SEM). Photospectrometrically the cells' viability (MTT) and alkaline phosphatase (ALP) production were assessed. The transcription of osteoblast-specific markers was elucidated with polymerase chain reaction (PCR) tests. Cells on CS-containing scaffolds in the presence of dexamethasone showed the highest ALP production. PCR monitored an increase of osteoblastic markers. All scaffolds showed higher calcium deposition than cell-free controls. These results lead to the conclusion that a niche containing CS renders the differentiation of hMSCs toward osteoblastic cells more specific.
Collapse
|
34
|
Abstract
Radiation rapidly and persistently alters the soluble and insoluble components of the tissue microenvironment. This affects the cell phenotype, tissue composition and the physical interactions and signalling between cells. These alterations in the microenvironment can contribute to carcinogenesis and alter the tissue response to anticancer therapy. Examples of these responses and their implications are discussed with a view to therapeutic intervention.
Collapse
Affiliation(s)
- Mary Helen Barcellos-Hoff
- Life Sciences Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, California 94720, USA.
| | | | | |
Collapse
|
35
|
Han B, Liu J, Ma MJ, Zhao L. Clinicopathological significance of heparanase and basic fibroblast growth factor expression in human esophageal cancer. World J Gastroenterol 2005; 11:2188-92. [PMID: 15810091 PMCID: PMC4305794 DOI: 10.3748/wjg.v11.i14.2188] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: Human heparanase is an endo-D-glucuronidase that degrades heparan sulfate/heparin and has been implicated in a variety of biological processes. The objective was to investigate the expression of heparanase (Hps) and basic fibroblast growth factor (bFGF) and their relationship to neoangiogenesis and metastasis of human esophageal carcinoma.
METHODS: Seventy-nine patients who had undergone esophageal resection for esophageal carcinoma without preoperative treatment were included in the present study. Immunohistochemistry was used to study the expression of Hps, bFGF and microvessel density (MVD) in 79 cases of esoph-ageal carcinoma. bFGF and Hps were quantitatively detected with immunohistochemistry in 79 cases of human esopha-geal carcinoma and 19 cases of adjacent normal human esophageal carcinoma. Cd34 was used to explore the MVD as a marker of endothelial cells.
RESULTS: Hps and bFGF expression in tumor tissue, being remarkably higher than that in normal esophageal tissue, were significantly correlated with clinicopathological features (depth of invasion, lymph-node metastasis and TNM stage) and MVD.
CONCLUSION: The results of this study suggest that the coexpression of Hps and bFGF plays a key role in angiogenesis, invasion and metastasis of esophageal carcinoma. Hps and bFGF may serve as a predictor of progression in esophageal carcinoma. The expression of heparanase in esophageal carcinoma enhances growth, invasion, and angiogenesis of the tumor, and bFGF seems to be a potent antigenic factor for esophageal carcinoma.
Collapse
Affiliation(s)
- Biao Han
- First Affiliated Hospital, Lanzhou, Medical College, Lanzhou 730000, Gansu Province, China
| | | | | | | |
Collapse
|
36
|
Chen Y, Ali T, Todorovic V, O'leary JM, Kristina Downing A, Rifkin DB. Amino acid requirements for formation of the TGF-beta-latent TGF-beta binding protein complexes. J Mol Biol 2005; 345:175-86. [PMID: 15567420 DOI: 10.1016/j.jmb.2004.10.039] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2004] [Accepted: 10/14/2004] [Indexed: 01/11/2023]
Abstract
Transforming growth factor beta (TGF-beta) is secreted primarily as a latent complex consisting of the TGF-beta homodimer, the TGF-beta propeptides (called the latency-associated protein or LAP) and the latent TGF-beta binding protein (LTBP). Mature TGF-beta remains associated with LAP by non-covalent interactions that block TGF-beta from binding to its receptor. Complex formation between LAP and LTBP is mediated by an intramolecular disulfide exchange between the third 8-cysteine (8-Cys3) domain of LTBP with a pair of cysteine residues in LAP. Only the third 8-Cys domains of LTBP-1, -3, and -4 bind LAP. From comparison of the 8-Cys3(LTBP-1) structure with that of the non-TGF-beta-binding 8-Cys6(fibrillin-1), we observed that a two-residue insertion in 8-Cys3(LTBP-1) increased the potential for disulfide exchange of the 2-6 disulfide bond. We further proposed that five negatively charged amino acid residues surrounding this bond mediate initial protein-protein association. To validate this hypothesis, we monitored binding by fluorescence resonance energy transfer (FRET) analysis and co-expression assays with TGF-beta1 LAP (LAP-1) and wild-type and mutant 8-Cys3 domains. FRET experiments demonstrated ionic interactions between LAP-1 and 8-Cys3. Mutation of the five amino acid residues revealed that efficient complex formation is most dependent on two of these residues. Although 8-Cys3(LTBP-1) binds proTGF-betas effectively, the domain from LTBP-4 does so poorly. We speculated that this difference was due to the substitution of three acidic residues by alanine, serine, and arginine in the LTBP-4 sequence. Additional experiments with 8-Cys3(LTBP-4) indicated that enhanced binding of LAP to 8-Cys3(LTBP-4) is achieved if the residues A, S, and R are changed to those in 8-Cys3(LTBP1) (D, D, and E) and the QQ dipeptide insertion of LTBP-4 is changed to the FP in 8-Cys3(LTBP-1). These studies identify surface residues that contribute to the interactions of 8-Cys3 and LAP-1 and may yield information germane to the interaction of 8-Cys domains and additional TGF-beta superfamily propeptides, an emerging paradigm for growth factor regulation.
Collapse
Affiliation(s)
- Yan Chen
- Department of Cell Biology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | | | | | | | | | | |
Collapse
|
37
|
Smith MF, Gutierrez CG, Ricke WA, Armstrong DG, Webb R. Production of matrix metalloproteinases by cultured bovine theca and granulosa cells. Reproduction 2005; 129:75-87. [PMID: 15615900 DOI: 10.1530/rep.1.00381] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Matrix metalloproteinases (MMPs) degrade the proteinaceous components of the extracellular matrix and are presumably essential for follicular growth culminating in ovulation or atresia. The objectives of this study were to characterize the gelatinolytic and caseinolytic MMPs secreted by cultured bovine thecal and granulosal cells and to determine the effect of luteinizing hormone (LH) on MMP secretion. Thecal and granulosal cells were collected from small bovine follicles (<5 mm) on day 2 or 5 of the estrous cycle (day 0 = estrus). A serum-free culture system was utilized in which bovine thecal and granulosal cells do not spontaneously luteinize, but produce androstenedione and estradiol in response to physiological concentrations of LH and follicle-stimulating hormone (FSH) respectively. The effect of LH (0, 1 or 100 ng/ml) on MMP production was determined in conditioned media collected every 48 h for 144 h. MMPs were detected by gelatin and casein zymography and MMP activity was quantified by image analysis. Thecal and granulosal cell conditioned media contained MMPs that had a relative molecular size (Mr) ranging from 53 000 to 200 000 and addition of 1,10 phenanthroline (MMP inhibitor) blocked gelatinolytic and caseinolytic activity. Patterns of gelatinolytic activity in thecal and granulosal cell conditioned media differed over time with theMr62 000 and 83 000 MMPs being increased (P< 0.05) and theMr53 000 MMP being decreased (P< 0.05) at 96 h of culture. LH (1 or 100 ng/ml) increased (P< 0.05) gelatinolytic activity of theMr53 000 and 62 000 gelatinases within thecal cell conditioned media but not granulosal cell conditioned media. TheMr62 000 and 83 000 gelatinolytic activities corresponded to the active forms of gelatinase A (Mr62 000) and B (Mr, 83 000) and gelatinase A was detected in thecal cell conditioned media by Western blot analysis. Caseinolytic activity (Mr83 000) was detected in both thecal and granulosal cell conditioned media and increased from 48 to 96 h. In summary, thecal and granulosal cells secrete gelatinolytic and caseinolytic MMPs and thecal cell production of gelatinase A was stimulated by LH.
Collapse
Affiliation(s)
- M F Smith
- Department of Animal Science, University of Missouri-Columbia, Columbia MO 65211, USA.
| | | | | | | | | |
Collapse
|
38
|
Annes JP, Chen Y, Munger JS, Rifkin DB. Integrin alphaVbeta6-mediated activation of latent TGF-beta requires the latent TGF-beta binding protein-1. ACTA ACUST UNITED AC 2004; 165:723-34. [PMID: 15184403 PMCID: PMC2172370 DOI: 10.1083/jcb.200312172] [Citation(s) in RCA: 390] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Transforming growth factor-βs (TGF-β) are secreted as inactive complexes containing the TGF-β, the TGF-β propeptide, also called the latency-associated protein (LAP), and the latent TGF-β binding protein (LTBP). Extracellular activation of this complex is a critical but incompletely understood step in TGF-β regulation. We have investigated the role of LTBP in modulating TGF-β generation by the integrin αVβ6. We show that even though αvβ6 recognizes an RGD on LAP, LTBP-1 is required for αVβ6-mediated latent TGF-β activation. The domains of LTBP-1 necessary for activation include the TGF-β propeptide-binding domain and a basic amino acid sequence (hinge domain) with ECM targeting properties. Our results demonstrate an LTBP-1 isoform-specific function in αVβ6-mediated latent TGF-β activation; LTBP-3 is unable to substitute for LTBP-1 in this assay. The results reveal a functional role for LTBP-1 in latent TGF-β activation and suggest that activation of specific latent complexes is regulated by distinct mechanisms that may be determined by the LTBP isoform and its potential interaction with the matrix.
Collapse
Affiliation(s)
- Justin P Annes
- Department of Cell Biology, New York University School of Medicine, 550 First Ave., New York, NY 10016, USA
| | | | | | | |
Collapse
|
39
|
Garcia CM, Darland DC, Massingham LJ, D'Amore PA. Endothelial cell-astrocyte interactions and TGF beta are required for induction of blood-neural barrier properties. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2004; 152:25-38. [PMID: 15283992 DOI: 10.1016/j.devbrainres.2004.05.008] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/16/2004] [Indexed: 11/25/2022]
Abstract
We sought to establish a blood-neural barrier (BNB) model of astrocyte contact with endothelial cells (EC) to test the hypothesis that transforming growth factor beta (TGF beta) promotes an EC barrier-phenotype. Astrocyte-EC contact induced BNB properties in EC. Transendothelial resistance was augmented by direct contact between astrocytes-EC, but not by astrocyte-conditioned medium or astrocyte-EC coculture conditioned medium. Coculture of EC and astrocytes led to significant increase in endothelial occludin levels and junctional localization. EC gamma-glutamyl-transferase (GGT) activity was increased by direct contact with astrocytes, by conditioned medium from cocultures or by TGF beta1. Coculture inhibited EC proliferation with no effect on astrocyte proliferation. A neutralizing antibody to TGF beta decreased GGT activity in cocultures and increased cell number. Whereas total TGF beta was not significantly altered by coculture, activated TGF beta increased in astrocyte-EC cocultures. In summary, astrocyte-EC contact induces BNB characteristics in EC and locally activated TGF beta is responsible for part of the induction.
Collapse
Affiliation(s)
- Claudia M Garcia
- Biological and Biomedical Sciences Graduate Program, Harvard Medical School, Boston, MA, USA
| | | | | | | |
Collapse
|
40
|
Lyon CJ, Hsueh WA. Effect of plasminogen activator inhibitor-1 in diabetes mellitus and cardiovascular disease. Am J Med 2003; 115 Suppl 8A:62S-68S. [PMID: 14678868 DOI: 10.1016/j.amjmed.2003.08.014] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Concentrations of plasminogen activator inhibitor-1 (PAI-1) are elevated beginning at the stage of impaired glucose tolerance and continuing through the development of diabetes mellitus and the metabolic syndrome. Evolving evidence of the central role of PAI-1 in mediating fibrosis and thrombosis increasingly supports the theory that it is a significant risk factor for macrovascular complications and cardiovascular disease, particularly in patients with diabetes. Several clinical studies have demonstrated a strong correlation between circulating PAI-1 levels and cardiovascular events and mortality. With the potentially severe effects of elevated PAI-1 levels becoming evident, there is increased interest in developing therapies targeted at reducing PAI-1 expression or circulating concentrations. Thus far, weight loss, inhibitors of the renin-angiotensin system, and insulin sensitization through use of thiazolidinediones (TZDs) appear to be the most promising strategies for managing elevated PAI-1 levels. Of these, TZD therapy is the only one that provides the benefits of both long-term glycemic control and improved cardiovascular risk profile. This article reviews the regulation of PAI-1, its activity in various disease states, and available treatment options.
Collapse
Affiliation(s)
- Christopher J Lyon
- Division of Endocrinology, Diabetes and Hypertension, University of California at Los Angeles, Los Angeles, California 90095, USA
| | | |
Collapse
|
41
|
Abstract
Cellular activities are primarily initiated, modulated and sustained by multifunctional molecules (cytokines and growth factors) that are secreted into the extracellular space and that signal through membrane-bound, high-affinity receptors. In contrast to the fairly well understood mechanisms that mediate the specificity of signal transduction within the confined and compartmentalized environment of the cell, significantly less is known about the mechanisms that regulate the availability of signaling molecules in the extracellular milieu. Recent findings have implicated the participation of extracellular protein macroaggregates in signaling events controlling patterning and morphogenesis. The results suggest a functional coupling between the tissue-specific organization of collagenous and elastic macroaggregates and their ability to perform instructive as well as structural functions. These observations open the way to a novel understanding in these poorly understood and critically important areas of cell and developmental biology.
Collapse
Affiliation(s)
- Francesco Ramirez
- Laboratory of Genetics and Organogenesis Hospital for Special Surgery, Weill Medical College of Cornell University, 535 East 70th Street, New York, NY 10021, USA.
| | | |
Collapse
|
42
|
Ewan KB, Shyamala G, Ravani SA, Tang Y, Akhurst R, Wakefield L, Barcellos-Hoff MH. Latent transforming growth factor-beta activation in mammary gland: regulation by ovarian hormones affects ductal and alveolar proliferation. THE AMERICAN JOURNAL OF PATHOLOGY 2002; 160:2081-93. [PMID: 12057913 PMCID: PMC1850834 DOI: 10.1016/s0002-9440(10)61158-3] [Citation(s) in RCA: 121] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/08/2002] [Indexed: 11/25/2022]
Abstract
Transforming growth factor-beta1 (TGF-beta 1) is a pluripotent cytokine that can inhibit epithelial proliferation and induce apoptosis, but is also widely implicated in breast cancer progression. Understanding its biological action in mammary development is critical for understanding its role in cancer. TGF-beta 1 is produced as a latent complex that requires extracellular activation before receptor binding. To better understand the spatial and temporal regulation of its action during mammary gland development, we examined the pattern of activation in situ using antibodies selected to distinguish between latent and active TGF-beta. Activation was highly restricted. TGF-beta 1 activation was localized primarily to the epithelium, and within the epithelium it was restricted to luminal epithelial cells but absent from either cap or myoepithelial cells. Within the luminal epithelium, we noted a further restriction. During periods of proliferation (ie, puberty, estrus and pregnancy), which are stimulated by ovarian hormones, TGF-beta 1 activation decreased in some cells, consistent with preparation for proliferation. Paradoxically, other cells simultaneously increase TGF-beta 1 immunoreactivity, which suggests that TGF-beta 1 differentially restrains epithelial subpopulations from responding to hormonal signals to proliferate. These data suggest that endogenous TGF-beta 1 activation and thus activity are regulated by ovarian hormones. To determine the specific consequences of TGF-beta 1 activity, we manipulated TGF-beta 1 levels in vivo using Tgfbeta 1 knockout mice and undertook tissue recombination experiments with heterozygous tissue. In Tgfbeta 1 heterozygous mice, which have <10% wild-type levels of TGF-beta1, ductal development during puberty and alveolar development during pregnancy were accelerated, consistent with its role as a growth inhibitor. The proliferative index of Tgfbeta 1+/- epithelium was increased approximately twofold in quiescent tissue and fourfold in proliferating tissue but both ducts and alveoli were grossly and histologically normal. To test whether epithelial TGF-beta1 was critical to the proliferative phenotype, Tgfbeta 1+/+ and +/- epithelium were transplanted into +/+ mammary stroma. The outgrowth of Tgfbeta 1+/- epithelium was accelerated in wild-type hosts, indicating that the phenotype was intrinsic to the epithelium. Moreover, proliferation was 15-fold greater in Tgfbeta 1+/- than wild-type mice after ovariectomy and treatment with estrogen and progesterone, suggesting that TGF-beta 1 acts in an autocrine or juxtacrine manner to regulate epithelial proliferation. Together these data indicate that ovarian hormones regulate TGF-beta 1 activation, which in turn restricts proliferative response to hormone signaling.
Collapse
Affiliation(s)
- Kenneth B Ewan
- Life Sciences Division, Lawrence Berkeley National Laboratory, University of California, Berkeley, California 94720, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Smith MF, Ricke WA, Bakke LJ, Dow MPD, Smith GW. Ovarian tissue remodeling: role of matrix metalloproteinases and their inhibitors. Mol Cell Endocrinol 2002; 191:45-56. [PMID: 12044918 DOI: 10.1016/s0303-7207(02)00054-0] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Follicular formation, growth or atresia, and ovulation as well as luteal formation and subsequent regression are dependent upon cyclical remodeling of the extracellular matrix (ECM). The proteinaceous and nonproteinaceous components of the ECM provide the tissue specific, extracellular architecture to which cells attach. Furthermore, the ECM modulates cellular activities through cellular surface receptors and serves as a reservoir for specific growth factors, cytokines, and binding proteins. The ability of the ECM to direct the proliferation, differentiation and function of cells implicates ECM remodeling in normal ovarian function. Specific components of the ECM are cleaved by matrix metalloproteinases (MMPs) whose activities are specifically inhibited by tissue inhibitors of metalloproteinases (TIMPs). MMPs are zinc- and calcium-dependent enzymes that collectively degrade proteinaceous components of the ECM. Controlled turnover of ECM by MMPs and TIMPs may be essential for creating and (or) preserving microenvironments conducive to follicular and luteal function and is likely dependent upon the ratio of enzyme to inhibitor. To date, most studies have focused upon correlating ovarian expression of MMPs and TIMPs with various stages of the reproductive cycle. From these studies, many potential key regulators of ovarian ECM remodeling have been identified. This review presents evidence for the involvement of MMPs and TIMPs in ECM remodeling associated with follicular and luteal function.
Collapse
Affiliation(s)
- Michael F Smith
- Department of Animal Sciences, 160 Animal Science Research Center, University of Missouri, Columbia, MO 65211, USA.
| | | | | | | | | |
Collapse
|
44
|
Steffensen B, Häkkinen L, Larjava H. Proteolytic events of wound-healing--coordinated interactions among matrix metalloproteinases (MMPs), integrins, and extracellular matrix molecules. CRITICAL REVIEWS IN ORAL BIOLOGY AND MEDICINE : AN OFFICIAL PUBLICATION OF THE AMERICAN ASSOCIATION OF ORAL BIOLOGISTS 2002; 12:373-98. [PMID: 12002821 DOI: 10.1177/10454411010120050201] [Citation(s) in RCA: 160] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
During wound-healing, cells are required to migrate rapidly into the wound site via a proteolytically generated pathway in the provisional matrix, to produce new extracellular matrix, and, subsequently, to remodel the newly formed tissue matrix during the maturation phase. Two classes of molecules cooperate closely to achieve this goal, namely, the matrix adhesion and signaling receptors, the integrins, and matrix-degrading and -processing enzymes, the matrix metalloproteinases (MMPs). There is now substantial experimental evidence that blocking key molecules of either group will prevent or seriously delay wound-healing. It has been known for some time now that cell adhesion by means of the integrins regulates the expression of MMPs. In addition, certain MMPs can bind to integrins or other receptors on the cell surface involved in enzyme activation, thereby providing a mechanism for localized matrix degradation. By proteolytically modifying the existing matrix molecules, the MMPs can then induce changes in cell behavior and function from a state of rest to migration. During wound repair, the expression of integrins and MMPs is simultaneously up-regulated. This review will focus on those aspects of the extensive knowledge of fibroblast and keratinocyte MMPs and integrins in biological processes that relate to wound-healing.
Collapse
Affiliation(s)
- B Steffensen
- Department of Periodontics, University of Texas Health Science Center at San Antonio, 78229-3900, USA.
| | | | | |
Collapse
|
45
|
Northcott C, Florian JA, Dorrance A, Watts SW. Arterial epidermal growth factor receptor expression in deoxycorticosterone acetate-salt hypertension. Hypertension 2001; 38:1337-41. [PMID: 11751714 DOI: 10.1161/hy1201.096815] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Epidermal growth factor (EGF) causes contraction in arteries from deoxycorticosterone acetate (DOCA)-salt hypertensive rats but not in normotensive sham rats. We hypothesized that an increase in the number of EGF receptors (EGFRs) in arteries from DOCA-salt rats enables the observed contraction to EGF to occur. DOCA-salt rats had a systolic blood pressure >170 mm Hg, whereas all sham rats had a systolic blood pressure <125 mm Hg. Thoracic aorta were removed for measurement of isometric force, EGFR mRNA levels, and EGFR protein levels. EGF caused a significant contraction in endothelium-denuded aorta from DOCA-salt rats (38+/-7% of maximal phenylephrine-induced [10 micromol/L] contraction) compared with aorta from sham rats (4+/-2%). The EGFR tyrosine kinase-specific inhibitors 4,5-dianilinophthalimide (10 micromol/L) and AG1478 (250 nmol/L) reduced contraction in aorta from DOCA-salt by 85+/-14% and 65+/-10%, respectively. EGFR mRNA in DOCA-salt aorta was increased 4.2-fold compared with that in sham aorta. However, Western analyses of membrane-enriched and whole-tissue lysate of aorta from sham and DOCA-salt revealed no statistical difference in the density of EGFR protein between sham and DOCA-salt aorta. These data refute our hypothesis and suggest that a change downstream of EGFR is responsible for enabling EGF-induced contraction in hypertension.
Collapse
Affiliation(s)
- C Northcott
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing 48824-1317, USA
| | | | | | | |
Collapse
|
46
|
Affiliation(s)
- W S Yeo
- Department of Chemistry, University of Chicago, 5735 South Ellis Avenue Chicago, IL 60637, USA
| | | | | |
Collapse
|
47
|
Pepper MS. Role of the matrix metalloproteinase and plasminogen activator-plasmin systems in angiogenesis. Arterioscler Thromb Vasc Biol 2001; 21:1104-17. [PMID: 11451738 DOI: 10.1161/hq0701.093685] [Citation(s) in RCA: 529] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Extracellular proteolysis is an absolute requirement for new blood vessel formation (angiogenesis). This review examines the role of the matrix metalloproteinase (MMP) and plasminogen activator (PA)-plasmin systems during angiogenesis. Specifically, a role for gelatinases (MMP-2, MMP-9), membrane-type 1 MMP (MMP-14), the urokinase-type PA receptor, and PA inhibitor 1 has been clearly defined in a number of model systems. The MMP and PA-plasmin systems have also been implicated in experimental vascular tumor formation, and their role during this process will be examined. Antiproteolysis, particularly in the context of angiogenesis, has become a key target in therapeutic strategies aimed at inhibiting tumor growth and other diseases associated with neovascularization.
Collapse
Affiliation(s)
- M S Pepper
- Department of Morphology, University Medical Center, Geneva, Switzerland.
| |
Collapse
|
48
|
Minter RM, Ferry MA, Rectenwald JE, Bahjat FR, Oberholzer A, Oberholzer C, La Face D, Tsai V, Ahmed CM, Hutchins B, Copeland EM, Ginsberg HS, Moldawer LL. Extended lung expression and increased tissue localization of viral IL-10 with adenoviral gene therapy. Proc Natl Acad Sci U S A 2001; 98:277-82. [PMID: 11134537 PMCID: PMC14581 DOI: 10.1073/pnas.98.1.277] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
IL-10 is a pleiotropic cytokine that acts as an important regulator of macrophage, T cell, and natural killer cell functions. Human IL-10 (hIL-10) has both stimulatory and inhibitory effects on a wide variety of cell types. Viral IL-10 (vIL-10) possesses only a subset of hIL-10's activities, predominantly its suppression of cytokine synthesis by T helper type 1 clones. In the present report, we evaluated tissue accumulation and biological activity of hIL-10 and vIL-10 in vivo in individual organs by using a first-generation adenoviral (Ad) vector administered intratracheally and intravenously. We report the observation that Ad vectors delivering vIL-10, but not hIL-10, are associated with prolonged expression in the lung (>42 days) when delivered intratracheally. In contrast, there was no prolongation in vIL-10 expression when Ad vectors were intravenously administered, although vIL-10 levels in the tissue, but not serum, were markedly increased relative to hIL-10. Moreover, we report an augmented capacity of expressed vIL-10 versus hIL-10 to suppress the acute inflammatory responses in the lung to intratracheal administration of Ad. These findings confirm fundamental differences in Ad-induced expression of vIL-10 and hIL-10 when administered to the lungs. The results further suggest that Ad vectors expressing vIL-10 may have a role as anti-inflammatory agents in the treatment of acute and chronic lung inflammation.
Collapse
Affiliation(s)
- R M Minter
- Department of Surgery, University of Florida College of Medicine, P.O. Box 100286, Gainesville, FL 32610, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Extended lung expression and increased tissue localization of viral IL-10 with adenoviral gene therapy. Proc Natl Acad Sci U S A 2001; 98. [PMID: 11134537 PMCID: PMC14581 DOI: 10.1073/pnas.250489297] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
IL-10 is a pleiotropic cytokine that acts as an important regulator of macrophage, T cell, and natural killer cell functions. Human IL-10 (hIL-10) has both stimulatory and inhibitory effects on a wide variety of cell types. Viral IL-10 (vIL-10) possesses only a subset of hIL-10's activities, predominantly its suppression of cytokine synthesis by T helper type 1 clones. In the present report, we evaluated tissue accumulation and biological activity of hIL-10 and vIL-10 in vivo in individual organs by using a first-generation adenoviral (Ad) vector administered intratracheally and intravenously. We report the observation that Ad vectors delivering vIL-10, but not hIL-10, are associated with prolonged expression in the lung (>42 days) when delivered intratracheally. In contrast, there was no prolongation in vIL-10 expression when Ad vectors were intravenously administered, although vIL-10 levels in the tissue, but not serum, were markedly increased relative to hIL-10. Moreover, we report an augmented capacity of expressed vIL-10 versus hIL-10 to suppress the acute inflammatory responses in the lung to intratracheal administration of Ad. These findings confirm fundamental differences in Ad-induced expression of vIL-10 and hIL-10 when administered to the lungs. The results further suggest that Ad vectors expressing vIL-10 may have a role as anti-inflammatory agents in the treatment of acute and chronic lung inflammation.
Collapse
|
50
|
Wood TL, Rogler LE, Czick ME, Schuller AG, Pintar JE. Selective alterations in organ sizes in mice with a targeted disruption of the insulin-like growth factor binding protein-2 gene. Mol Endocrinol 2000; 14:1472-82. [PMID: 10976924 DOI: 10.1210/mend.14.9.0517] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Insulin-like growth factor binding protein 2 (IGFBP-2) is one member of the family of IGF binding proteins believed to have both endocrine functions elicited by modulating serum IGF half-life and transport as well as autocrine/paracrine functions that result from blocking or enhancing the availability of IGFs to bind cell surface receptors. To clarify the in vivo role of IGFBP-2, we have used gene targeting to introduce a null IGFBP-2 allele into the mouse genome. Animals homozygous for the altered allele are viable and fertile, contain no IGFBP-2 mRNA, and have no detectable IGFBP-2 in the adult circulation. Heterozygous and homozygous animals showed no significant differences in prenatal or postnatal body growth. Analyses of organ weights in adult males, however, revealed that spleen weight was reduced and liver weight was increased in the absence of IGFBP-2. In addition, ligand blot analyses of sera from adult IGFBP-2 null males showed that IGFBP-1, IGFBP-3, and IGFBP-4 levels were increased relative to wild-type mice. These results demonstrate that up-regulation of multiple IGFBPs accompanies the absence of IGFBP-2 and that IGFBP-2 has a critical role, either directly or indirectly, in modulating spleen and liver size.
Collapse
Affiliation(s)
- T L Wood
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, University of Medicine and Dentistry New Jersey, Piscataway 08854, USA
| | | | | | | | | |
Collapse
|